Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nutr Res ; 85: 99-118, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33460863

RESUMEN

An amorphous formula of curcumin (CUR) has shown to enable an improved bioavailability after ingestion. The aim of this study was to investigate the hypothesis that exogenously administered CUR has an advantage in ameliorating post-traumatic stress disorder at low doses. To this end, Long-Evans rats were dietary exposed to CUR at 0.1% or 0.5% from gestational day 6 to postnatal day (PND) 74 or 77. Offspring exposed to 0.1% CUR revealed facilitation of anti-anxiety-like behavior in the open field test and fear-extinction learning tested during PND 62 to 74, increases in hippocampal granule cells expressing immediate-early gene proteins and a decrease in prelimbic cortical neurons expressing phosphorylated extracellular signal-regulated kinase 1/2 after the last trial of the fear-extinction learning test on PND 74. The constitutive gene expression levels of Gria1, Gria2, Grin2d, Slc17a6, and Slc17a7 were altered in the hippocampal dentate gyrus and amygdala on PND 77. These results suggest alterations in synaptic plasticity to strengthen neural circuits in promoting the behavioral effects by 0.1%-CUR. In contrast, 0.5% CUR revealed a lack of any of the changes in behavioral tests that were observed at 0.1%; however, this dose upregulated oxidative stress and neuroinflammation-related genes in the hippocampal dentate gyrus, and increased neural stem cells and proliferation activity of the subgranular zone in the dentate gyrus. These results suggest a possible preventive use of CUR at low doses in mitigating some stress disorders; however, excessively absorbed doses may prevent behavioral changes by inducing neuroinflammation that affects hippocampal neurogenesis involving neural stem cells.


Asunto(s)
Ansiedad , Conducta Animal , Encéfalo/fisiología , Curcumina/administración & dosificación , Miedo , Animales , Animales Recién Nacidos , Condicionamiento Psicológico , Curcumina/análisis , Curcumina/farmacología , Giro Dentado/fisiología , Extinción Psicológica , Femenino , Expresión Génica , Hipocampo/fisiología , Masculino , Neurogénesis , Neuroglía/citología , Plasticidad Neuronal , Neuronas/metabolismo , Corteza Prefrontal/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Long-Evans , Sinapsis/fisiología
2.
J Toxicol Pathol ; 33(4): 247-263, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33239843

RESUMEN

We previously reported that exposure to α-glycosyl isoquercitrin (AGIQ) from the fetal stage to adulthood facilitated fear extinction learning in rats. The present study investigated the specific AGIQ exposure period sufficient for inducing this behavioral effect. Rats were dietarily exposed to 0.5% AGIQ from the postweaning stage to adulthood (PW-AGIQ), the fetal stage to postweaning stage (DEV-AGIQ), or the fetal stage to adulthood (WP-AGIQ). Fear memory, anxiety-like behavior, and object recognition memory were assessed during adulthood. Fear extinction learning was exclusively facilitated in the WP-AGIQ rats. Synaptic plasticity-related genes showed a similar pattern of constitutive expression changes in the hippocampal dentate gyrus and prelimbic medial prefrontal cortex (mPFC) between the DEV-AGIQ and WP-AGIQ rats. However, WP-AGIQ rats revealed more genes constitutively upregulated in the infralimbic mPFC and amygdala than DEV-AGIQ rats, as well as FOS-immunoreactive(+) neurons constitutively increased in the infralimbic cortex. Ninety minutes after the last fear extinction trial, many synaptic plasticity-related genes (encoding Ephs/Ephrins, glutamate receptors/transporters, and immediate-early gene proteins and their regulator, extracellular signal-regulated kinase 2 [ERK2]) were upregulated in the dentate gyrus and amygdala in WP-AGIQ rats. Additionally, WP-AGIQ rats exhibited increased phosphorylated ERK1/2+ neurons in both the prelimbic and infralimbic cortices. These results suggest that AGIQ exposure from the fetal stage to adulthood is necessary for facilitating fear extinction learning. Furthermore, constitutive and learning-dependent upregulation of synaptic plasticity-related genes/molecules may be differentially involved in brain regions that regulate fear memory. Thus, new learning-related neural circuits for facilitating fear extinction can be established in the mPFC.

3.
J Appl Toxicol ; 40(11): 1467-1479, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32596862

RESUMEN

We previously found downregulation of low-density lipoprotein receptor class A domain-containing protein 4 (LDLRAD4), a negative regulator of transforming growth factor (TGF)-ß signaling, in glutathione S-transferase placental form (GST-P) expressing (+ ) pre-neoplastic lesions produced by treatment with nongenotoxic hepatocarcinogens for up to 90 days in rats. Here, we investigated the relationship between LDLRAD4 downregulation and TGFß signaling in nongenotoxic hepatocarcinogenesis. The transcripts of Tgfb and Hb-egf increased after ≥28 days of treatment. After 84 or 90 days, Snai1 increased transcripts and the subpopulation of GST-P+ foci downregulating LDLRAD4 co-expressed TGFß1, phosphorylated EGFR, or phosphorylated AKT2, and downregulated PTEN, showing higher incidences than those in GST-P+ foci expressing LDLRAD4. The subpopulation of GST-P+ foci downregulating LDLRAD4 also co-expressed caveolin-1 or TACE/ADAM17, suggesting that disruptive activation of TGFß signaling through a loss of LDLRAD4 enhances EGFR and PTEN/AKT-dependent pathways via caveolin-1-dependent activation of TACE/ADAM17 during nongenotoxic hepatocarcinogenesis. The numbers of c-MYC+ cells and PCNA+ cells were higher in LDLRAD4-downregulated GST-P+ foci than in LDLRAD4-expressing GST-P+ foci, suggesting a preferential proliferation of pre-neoplastic cells by LDLRAD4 downregulation. Nongenotoxic hepatocarcinogens markedly downregulated Nox4 after 28 days and later decreased cleaved caspase 3+ cells in LDLRAD4-downregulated GST-P+ foci, suggesting an attenuation of apoptosis by LDLRAD4 downregulation through activation of the EGFR pathway. At the late hepatocarcinogenesis stage in a two-stage model, LDLRAD4 downregulation was higher in adenoma and carcinoma than in pre-neoplastic cell foci, suggesting a role of LDLRAD4 downregulation in tumor development. Our results suggest that nongenotoxic hepatocarcinogens cause disruptive activation of TGFß signaling through downregulating LDLRAD4 toward carcinogenesis in the rat liver.


Asunto(s)
Apoptosis , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/metabolismo , Lesiones Precancerosas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Tetracloruro de Carbono , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Dietilnitrosamina , Modelos Animales de Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Metapirileno , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Ratas Endogámicas F344 , Transducción de Señal , Tioacetamida , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética
4.
J Toxicol Pathol ; 32(4): 261-274, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31719753

RESUMEN

To clarify difference in the responses on the reprogramming of metabolism toward carcinogenesis between genotoxic and non-genotoxic hepatocarcinogens in the liver, rats were repeatedly administered genotoxic hepatocarcinogens (N-nitrosodiethylamine, aflatoxin B1, N-nitrosopyrrolidine, or carbadox) or non-genotoxic hepatocarcinogens (carbon tetrachloride, thioacetamide, or methapyrilene hydrochloride) for 28, 84, or 90 days. Non-genotoxic hepatocarcinogens revealed transcript expression changes suggestive of suppressed mitochondrial oxidative phosphorylation (OXPHOS) after 28 days and increased glutathione S-transferase placental form-positive (GST-P+) foci downregulating adenosine triphosphate (ATP) synthase subunit beta, mitochondrial precursor (ATPB), compared with genotoxic hepatocarcinogens after 84 or 90 days, suggesting that non-genotoxic hepatocarcinogens are prone to suppress OXPHOS from the early stage of treatment, which is in contrast to genotoxic hepatocarcinogens. Both genotoxic and non-genotoxic hepatocarcinogens upregulated glycolytic enzyme genes and increased cellular membrane solute carrier family 2, facilitated glucose transporter member 1 (GLUT1) expression in GST-P+ foci for up to 90 days, suggesting induction of a metabolic shift from OXPHOS to glycolysis at early hepatocarcinogenesis by hepatocarcinogens unrelated to genotoxic potential. Non-genotoxic hepatocarcinogens increased c-MYC+ cells after 28 days and downregulated Tp53 after 84 or 90 days, suggesting a commitment to enhanced metabolic shift and cell proliferation. Genotoxic hepatocarcinogens also enhanced c-MYC activation-related metabolic shift until 84 or 90 days. In addition, both genotoxic and non-genotoxic hepatocarcinogens upregulated glutaminolysis-related Slc1a5 or Gls, or both, after 28 days and induced liver cell foci immunoreactive for neutral amino acid transporter B(0) (SLC1A5) in the subpopulation of GST-P+ foci after 84 or 90 days, suggesting glutaminolysis-mediated facilitation of cell proliferation toward hepatocarcinogenesis. These results suggest differential responses between genotoxic and non-genotoxic hepatocarcinogens on reprogramming of energy metabolic pathways toward carcinogenesis in liver cells from the early stage of hepatocarcinogen treatment.

5.
J Toxicol Pathol ; 32(3): 165-180, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31404398

RESUMEN

Hypothyroidism during the developmental stage induces disruption of hippocampal neurogenesis in later life, as well as inducing oxidative stress in the brain. The present study investigated the preventive effect of co-exposure to an antioxidant on disruptive neurogenesis induced by developmental exposure to anti-thyroid agent in rats. For this purpose, we used two antioxidants, α-glycosyl isoquercitrin (AGIQ) and α-lipoic acid (ALA). Mated female Sprague Dawley rats were either untreated (control) or treated with 12 ppm 6-propyl-2-thiouracil (PTU), an anti-thyroid agent, in drinking water from gestational day 6 to postnatal day (PND) 21, the latter group being subjected to feeding basal diet alone or diet containing AGIQ at 5,000 ppm or ALA at 2,000 ppm during PTU exposure. On PND 21, PTU-exposed offspring showed reductions in a broad range of granule cell lineage subpopulations and a change in the number of GABAergic interneuron subpopulations. Co-exposure of AGIQ or ALA with PTU altered the transcript levels of many genes across multiple functions, suggestive of enhancement of synaptic plasticity and neurogenesis. Nevertheless, immunohistochemical results did not support these changes. PTU exposure and co-exposure of AGIQ or ALA with PTU did not alter the hippocampal lipid peroxidation level. The obtained results suggest a possibility that thyroid hormone depletion itself primarily disrupts neurogenesis and that oxidative stress may not be involved in the disruption during development. Transcript expression changes of many genes caused by antioxidants may be the result of neuroprotective actions of antioxidants rather than their antioxidant activity. However, no preventive effect on neurogenesis suggested impairment of protein synthesis via an effect on mRNA translation due to hypothyroidism.

6.
J Toxicol Sci ; 44(5): 357-372, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31068541

RESUMEN

Developmental hypothyroidism as a model of autism spectrum disorders disrupts hippocampal neurogenesis through the adult stage. The present study investigated the ameliorating effect of postweaning exposure to antioxidant on the hypothyroidism-induced disruptive neurogenesis. Mated female Sprague-Dawley rats were treated with 0 or 10 ppm 6-propyl-2-thiouracil (PTU) as an anti-thyroid agent in drinking water from gestational day 6 to postnatal day (PND) 21 on weaning. PTU-exposed male offspring were fed either basal diet, diet containing α-glycosyl isoquercitrin (AGIQ) at 5,000 ppm or α-lipoic acid (ALA) at 1,000 ppm as an antioxidant from PND 21 to PND 77. PTU-exposure decreased DCX+ and NeuN+ granule cell lineage subpopulations, synaptic plasticity-related FOS+ granule cells, and hilar PVALB+ and GAD67+ GABAergic interneurons, increased hilar SST+ and CALB2+ interneurons, and upregulated Gria3, Otx2, and antioxidant enzyme genes in the dentate gyrus on PND 77. These results suggest disruption of neurogenesis remained in relation with increase of oxidative stress and compensatory responses to the disruption at the adult stage. AGIQ recovered expression of some antioxidant enzyme genes and was effective for restoration of NeuN+ postmitotic granule cells and PVALB+ and SST+ interneurons. In contrast, ALA was effective for restoration of all interneuron subpopulations, as well as postmitotic granule cells, and upregulated Grin2a that may play a role for the restoration. Both antioxidants recovered expression of Otx2 and AGIQ-alone recovered Gria3, suggesting a reversal of disruptive neurogenesis by compensatory responses. Thus, postweaning antioxidant exposure may be effective for ameliorating developmental hypothyroidism-induced disruptive neurogenesis by restoring the function of regulatory system.


Asunto(s)
Antioxidantes/farmacología , Hipocampo/efectos de los fármacos , Hipotiroidismo/fisiopatología , Neurogénesis/efectos de los fármacos , Quercetina/análogos & derivados , Ácido Tióctico/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteína Doblecortina , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Hipotiroidismo/inducido químicamente , Hipotiroidismo/genética , Masculino , Embarazo , Propiltiouracilo , Quercetina/farmacología , Ratas Sprague-Dawley
7.
J Toxicol Sci ; 44(2): 93-105, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30726815

RESUMEN

Maternal exposure to 3,3'-iminodipropionitrile (IDPN) affects hippocampal neurogenesis in mouse offspring, with biphasic disruption, which facilitates neurogenesis during exposure and reduces the broad range of the granule cell lineage population at the adult stage. The present study investigated the epigenetically hypermethylated and downregulated genes related to the IDPN-induced disrupted neurogenesis. Mated female mice were treated with IDPN at 0 or 1200 ppm in drinking water from gestational day 6 to postnatal day (PND) 21 on weaning. The hippocampal dentate gyrus of male offspring on PND 21 was subjected to methyl-capture sequencing and real-time reverse transcription-PCR analyses, followed by validation analyses on DNA methylation. Three genes, Edc4, Kiss1 and Mrpl38, were identified as those showing promoter-region hypermethylation and transcript downregulation, with Mrpl38 sustaining the changes through PND 77. Immunohistochemically, MRPL38, a mitochondrial ribosomal protein, revealed an irreversible decrease in the number of immunoreactive interneurons in the dentate gyrus hilar region, suggesting a causal relationship with the long-lasting effect on neurogenesis by the impaired migration due to mitochondrial dysfunction of interneurons, which regulate the differentiation and survival of granule cell lineages. Downregulation of Edc4 may also be responsible for decreased neurogenesis on PND 77 owing to a mechanism involving interleukin-6 downregulation via processing body dysfunction. Downregulation of Kiss1 may be responsible for the facilitation of neurogenesis during IDPN-exposure due to decreased glutamatergic neurotransmission and also for suppressed neurogenesis on PND 77 due to decreased expression of immediate-early genes, which play a crucial role in the maintenance of cell differentiation or plasticity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Intercambio Materno-Fetal , Neurogénesis/efectos de los fármacos , Neurotoxinas/toxicidad , Nitrilos/toxicidad , Efectos Tardíos de la Exposición Prenatal , Animales , Metilación de ADN , Epigénesis Genética , Femenino , Hipocampo/fisiología , Kisspeptinas/genética , Masculino , Ratones Endogámicos ICR , Embarazo
8.
Toxicol Sci ; 169(1): 122-136, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30690589

RESUMEN

This study examined hypermethylated and downregulated genes specific to carbon tetrachloride (CCl4) by Methyl-Seq analysis combined with expression microarray analysis in the liver of rats treated with CCl4 or N-nitrosodiethylamine (DEN) for 28 days, by excluding those with DEN. Among 52 genes, Ldlrad4, Proc, Cdh17, and Nfia were confirmed to show promoter-region hypermethylation by methylation-specific quantitative PCR analysis on day 28. The transcript levels of these 4 genes decreased by real-time reverse transcription-PCR analysis in the livers of rats treated with nongenotoxic hepatocarcinogens for up to 90 days compared with untreated controls and genotoxic hepatocarcinogens. Immunohistochemically, LDLRAD4 and PROC showed decreased immunoreactivity, forming negative foci, in glutathione S-transferase placental form (GST-P)+ foci, and incidences of LDLRAD4- and PROC- foci in GST-P+ foci induced by treatment with nongenotoxic hepatocarcinogens for 84 or 90 days were increased compared with those with genotoxic hepatocarcinogens. In contrast, CDH17 and NFIA responded to hepatocarcinogens without any relation to the genotoxic potential of carcinogens. All 4 genes did not respond to renal carcinogens after treatment for 28 days. Considering that Ldlrad4 is a negative regulator of transforming growth factor-ß signaling, Proc participating in p21WAF1/CIP1 upregulation by activation, Cdh17 inducing cell cycle arrest by gene knockdown, and Nfia playing a role in a tumor-suppressor, all these genes may be potential in vivo epigenetic markers of nongenotoxic hepatocarcinogens from the early stages of treatment in terms of gene expression changes. LDLRAD4 and PROC may have a role in the development of preneoplastic lesions produced by nongenotoxic hepatocarcinogens.


Asunto(s)
Tetracloruro de Carbono/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Metilación de ADN/efectos de los fármacos , Dietilnitrosamina/toxicidad , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Hígado/efectos de los fármacos , Lesiones Precancerosas/inducido químicamente , Animales , Cadherinas/genética , Cadherinas/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación hacia Abajo , Gutatión-S-Transferasa pi/genética , Gutatión-S-Transferasa pi/metabolismo , Hígado/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteína C/genética , Proteína C/metabolismo , Ratas Endogámicas F344 , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Tiempo
9.
Neurotox Res ; 35(3): 668-683, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30488313

RESUMEN

We previously reported that developmental exposure to T-2 toxin caused transient disruption of the hippocampal neurogenesis targeting neural stem cells (NSCs) and early-stage progenitor cells involving oxidative stress on weaning in mouse offspring. The present study examined metallothionein (MT) expression changes and their cellular identity in brain regions of these animals. T-2 toxin at 0, 1, 3, and 9 mg/kg was given in the diet of maternal mice from gestational day 6 to postnatal day (PND) 21 on weaning. Offspring were maintained through PND 77 without T-2 toxin exposure. Male offspring were analyzed. Immunohistochemically, MT-I/II+ cells increased in the subgranular zone (SGZ) of the dentate gyrus and cerebral cortex at ≥ 3 mg/kg and in the hilus of the dentate gyrus, corpus callosum, and cerebellum at 9 mg/kg on PND 21, suggestive of operation of cytoprotective function against oxidative stress throughout the brain. Double immunohistochemistry analysis revealed MT-I/II+ SGZ cells to be NSCs and MT-I/II+ cells in other brain regions to be astrocytes as toxicity targets of T-2 toxin. Phosphorylated STAT3+ cell numbers increased only in the cerebellum in parallel with the increase of GFAP+ astrocytes at 9 mg/kg, suggesting a STAT3-mediated transcriptional GFAP upregulation in cerebellar astrocytes. In the dentate gyrus, Il1a, Il1r1, and Mt2 increased transcripts at 9 mg/kg, suggesting activation of the IL-1 signaling cascade, possibly causing MT-II upregulation. The increase of MT-I/II+ cells in all brain regions disappeared or was suppressed below the control level on PND 77, suggesting a recovery from the T-2 toxin-induced oxidative stress.


Asunto(s)
Astrocitos/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Metalotioneína/metabolismo , Células-Madre Neurales/efectos de los fármacos , Toxina T-2/efectos adversos , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Astrocitos/patología , Relación Dosis-Respuesta a Droga , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Ratones Endogámicos ICR , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Distribución Aleatoria
10.
Food Chem Toxicol ; 120: 590-602, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30071239

RESUMEN

To investigate the developmental exposure effect of citreoviridin (CIT) on postnatal hippocampal neurogenesis, pregnant ICR mice were dietary exposed to CIT at 0, 1, 3 and 10 ppm from gestation day 6 to postnatal day (PND) 21 on weaning. Offspring were maintained through PND 77 without CIT exposure. Male offspring were analyzed. At 10 ppm on PND 21, weak changes suggestive of neural stem cell reduction and progenitor cell proliferation were observed. Number of hilar CALB1+ interneurons reduced, suggesting an influence on neurogenesis. In contrast, number of hilar SST+ interneurons increased and Bdnf and Ntrk2 transcripts upregulated in the dentate gyrus, suggesting a facilitation of BDNF-TRKB signaling for progenitor cell proliferation. Transcript expression changes of an outside regulatory system suggested suppressed function of GABAergic interneurons, especially of PVALB+ interneurons for compensation on neural stem cell reduction. At ≥ 3 ppm, number of ARC+ mature granule cells increased, and at 10 ppm, number of hilar GRIA1+ cells increased and Gria2 and Gria3 upregulated, suggesting an operation of AMPA receptor membrane trafficking on the increase of ARC-mediated synaptic plasticity. On PND 77, all the transcript expression changes of the neurogenesis regulatory system except for Grin2d were inverted, suggesting an operation of a homeostatic mechanism on CIT-induced disruptive neurogenesis. Simultaneous downregulation of Grin2a and Grin2d suggests suppression of GABAergic interneuron function to adjust neurogenesis at the normal level. The no-observed-adverse-effect level of CIT for offspring neurogenesis was determined to be 1 ppm, translating to 0.13-0.51 mg/kg body weight/day of maternal oral exposure.


Asunto(s)
Aurovertinas/toxicidad , Hipocampo/efectos de los fármacos , Micotoxinas/toxicidad , Neurogénesis/efectos de los fármacos , Neurotoxinas/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Apoptosis , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proliferación Celular , Relación Dosis-Respuesta a Droga , Femenino , Contaminación de Alimentos , Ácido Glutámico/metabolismo , Hipocampo/citología , Hipocampo/embriología , Hipocampo/metabolismo , Interneuronas/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Oryza/microbiología , Embarazo , Proteínas Tirosina Quinasas/metabolismo , Receptores AMPA/metabolismo , Transducción de Señal , Destete , Ácido gamma-Aminobutírico/metabolismo
11.
Toxicol Appl Pharmacol ; 334: 207-216, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28899750

RESUMEN

We previously found downregulation of ubiquitin-conjugating enzyme E2E 2 (UBE2E2) in GST-P-positive (+) proliferative lesions produced by tumor promotion from early hepatocarcinogenesis stages in rats. Here we investigated the role of UBE2E2 downregulation in preneoplastic lesions of the liver and other target organs produced by tumor promotion in rats. Increased number of UBE2E2-related ubiquitination target proteins, phosphorylated c-MYC, KDM4A and KMT5A, was found in the UBE2E2-downregulated GST-P+ foci, compared with GST-P+ foci expressing UBE2E2. However, p21WAF1/CIP1, another UBE2E2 target protein, did not increase in the positive cells. Furthermore, the numbers of PCNA+ cells and γH2AX+ cells were increased in UBE2E2-downregulated foci. These results suggest sustained activation of c-MYC and stabilization of KMT5A to result in c-MYC-mediated transcript upregulation and following KMT5A-mediated protein stabilization of PCNA in GST-P+ foci, as well as KDM4A stabilization resulting in slowing down of DNA damage response in these lesions. Similar results were also observed in GST-P+ foci produced by repeated treatment of rats with a hepatocarcinogen, thioacetamide, for 90days. Hepatocarcinogen treatment for 28 or 90days also increased the numbers of liver cells expressing UBE2E2-related ubiquitination target proteins, as well as PCNA+ or γH2AX+ cells. Conversely, UBE2E2 downregulation was lacking in PPARα agonist-induced hepatocarcinogenesis, as well as in carcinogenic processes targeting other organs, suggestive of the loss of UBE2E2-related ubiquitination limited to hepatocarcinogenesis producing GST-P+ proliferative lesions. Our results suggest that repeated hepatocarcinogen treatment of rats causes stabilization of UBE2E2-related ubiquitination target proteins in liver cells to promote carcinogenesis.


Asunto(s)
Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Gutatión-S-Transferasa pi/metabolismo , Hepatocitos/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Reparación del ADN , Regulación hacia Abajo , Regulación de la Expresión Génica , Gutatión-S-Transferasa pi/genética , Lesiones Precancerosas/inducido químicamente , Distribución Aleatoria , Ratas , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...